Вы находитесь на странице: 1из 17

,

Mechanism of degradation of the steroid side


chain in the formation of bile acids
Ingemar Bjorkhem
Department of Clinical Chemistry, Karolinska Institute, Huddinge Hospital, Huddinge, Sweden

Introduction in the steroid side-chain degradation convert the high-


The 27-Hydroxylase Pathway ly nonpolar Cpsteroid side-chain into a chain-shor-
27-Hydroxylation tened carboxylic acid conjugated to an amino acid.
Conversion of 27-hydroxysteroid to 27carboxysteroid
Peroxisomal Boxidation of coprostanoic acids These enzymes are mainly located in the mitochondria
The 25Hydroxylase Pathway and in the peroxisomes.
Relative Importance of the 25- and 27-Hydroxylase Pathways According to current concepts, the conversion of
Species Differences and Alternative Substrates for the cholesterol into bile acids in mammals starts with the

Downloaded from www.jlr.org by guest, on May 14, 2019


27-Hydroxylase Pathway nuclear transformations, and most or all of these chan-
Pathway involving 7a,27-dihydroxy-4-cholesten-3-oneas
intermediate ges precede those of the steroid side-chain. Based on
Pathway involving 7a,12a,27-trihydroxy4cholesten-3-one early in vivo and in vitro work on rats, the sequence of
as intermediate reactions shown in Fig. 1 was formulated about 25
Pathway involving 27-hydroxycholesteroI as intermediate years ago for the conversion of cholesterol into cholic
Inborn Errors in Side-Chain Cleavage and chenodeoxycholic acids. It is evident, however,
Cerebrotendinous xanthomatosis
Peroxisomal disorders that alternative pathways exist where some or most of
Mechanism of Degradation of the Steroid Side Chain of the changes in the steroid side-chain precede the
Plant Steroids changes in the nucleus. As the flux of bile acids
Concluding Remarks probably does not regulate any of the pathways where
7a-hydroxylation is not the first step, it seems that the
cholesterol 7a-hydroxylase is the only enzyme that is
INTRODUCTION capable of regulating the overall conversion into bile
acids.
The most important pathway for the metabolism The steroid side chain of plant sterols contains a
and excretion of cholesterol in mammals is the forma- methyl or an ethyl group in 24position. This means
tion of bile acids. The two major primary bile acids, that the normal mechanism for side-chain degradation
cholic and chenodeoxycholic acids, are formed in the cannot be operative. In view of the very low degree of
liver and secreted in bile to the intestine. absorption of these steroids, there is less need for a
The conversion of cholesterol into bile acids invol- specialized degradative enzyme. In at least one mam-
ves almost all the conceivable mechanisms for conver- malian species, however, a system has been evolved
sion of a lipophilic compound into an excretable that is able to convert plant sterols into highly polar
water-soluble product (for a general review, see ref. 1). conjugated Cnl-bile acids.
Of the more than 15 different enzymes participating In the present review, the emphasis is on the most
in the conversion, hydroxylases, oxidoreductases, and important mechanism for steroid side-chain degrada-
conjugating systems are of particular importance for tion in connection with bile acid biosynthesis, the 27-
increasing the polarity. The enzymes that modify the
steroid nucleus are able to convert the nonpolar
3~hydroxyA5-steroidinto a considerably more polar
5~holestane-?i~~,7~~-dihydroxy- or 5pcholestane-Sa,
7a,12a-trihydroxy steroid. The enzymes involved in Abbreviations: THCA, trihydroxycoprostanoic acid; DHCA,
these conversions are mainly located in the endoplas- dihydroxy-5~holestanoicacid (dihydroxycoprostanoic acid) ; CTX,
mic reticulum and the cytosol. The enzymes involved cerebrotendinous xanthomatosis.

Journal of Lipid Research Volume 33, 1992 455


HO
i
I
t '0H

0ai? 0
1

Downloaded from www.jlr.org by guest, on May 14, 2019


HO' HO'

CH,OH
t
CH20H

HO
HO'
I 1

H0'
+ HO'Z C O O H

HO'&OoH
HO' zoo.
Fig. 1. Major pathway for the normal biosynthesis of bile acids in the rat.

hydroxylase pathway. An alternative pathway that THE 27-HYDROXYLASE PATHWAY


operates when the major pathway is blocked will also
be reviewed. Alternative substrates for sidechain It is well established that the major mechanism for
degradation in different species, as well as inborn er- degradation of the cholesterol sidechain starts with
rors of metabolism affecting side-chain degradation, oxidation of one of the two terminal methyl groups
are also discussed. A recently discovered new pathway (ohydroxylation) followed by fhxidation and con-
for degradation of the steroid sidechain of plant jugation. A simplified scheme of the reactions is shown
sterols will be mentioned. in Fig. 2.

456 Journal of Lipid Research Volume 33, 1992


Downloaded from www.jlr.org by guest, on May 14, 2019
m R CO.CoA + <Co.CoA

. NHCH,COOH

HO’

Fig. 2. Side-chain oxidation and conjugation in connection with bile acid biosynthesis from cholesterol.
The 27-hydroxylase pathway for the conversion of 5&cholestane-3a,7a,12a-triol
into cholic acid is shown.

The above pathway will be referred to as the “W almost exclusively attacks the C-27 methyl group (for a
hydroxylase pathway” or the “27-hydroxylasepathway.” general review, see ref. 1 ) . This hydroxylase should
thus be denoted “27-hydroxylase,” although it has
27-Hydroxylation been referred to as “26hydroxylase” in most previous
Inasmuch as hydroxylation of one of the two ter- work. There is also a microsomal *hydroxylase in
minal methyl groups in the steroid sidechain creates mammalian liver, which has a specificity towards the C-
an asymetric carbon atom at C25, @hydroxylation 26 methyl group (3). In humans, this microsomal 2 6
may be stereospecific. Popjak et al. (2) have suggested hydroxylase has little activity compared to the
that the 25-pro-R methyl group should be denoted C- mitochondrial 27-hydroxylase (4).In accordance with
26 and the 25-pro-S methyl group C-27. There is a this, trihydroxycoprostanoic acid isolated from human
mitochondrial @hydroxylase in mammalian liver that bile has the 25-R-configuration (5), indicating that it is

Bj6rkhem Mechanism of degradation of steroid side chain in formation of bile acids 457
formed by an initial attack by the mitochondrial 27- showed that the substrate specificity of a reconstituted
hydroxylase. In addition to 26-hydroxylase activity, system from rat liver mitochondria was similar to that
both rat and human liver microsomes contain 2 3 , 2 4 , in intact mitochondria. The rate of 27-hydroxylation
and 25-hydroxylases active towards the Cnrsteroid side- was, however, about 10-fold higher in the reconstituted
chain (cf below). In the rat, the microsomal 26- system than in intact mitochondria, indicating that the
hydroxylase may be of importance and there is some transport of the steroid through the mitochondria
evidence that in this species the enzyme may be in- membranes may be a limiting factor (19).
volved in the regulation of the ratio between cholic Wikvall (20) and Dahlback and Wikvall (21) were
acid and chenodeoxycholic acid formed in the liver the first to purify the mitochondrial 27-hydroxylase to
(6). homogeneity. They used rabbit liver as enzyme source.
The microsomal 26-hydroxylase has a high substrate Later, other groups characterized the rat (22), human
specificity and the mitochondrial 27-hydroxylase has a (23), and pig (24) 27-hydroxylases. Andersson et al.
low substrate specificity (see below). The mitochon- (10) recently made an extensive characterization of
drial 27-hydroxylase is located not only in the liver but the rabbit liver 27-hydroxylase. The protein sequence
also in fibroblasts (7), brain (S), kidney (9), and of the 499 amino acid enzyme shares many of the
several other organs and tissues (10). hallmarks of a cytochrome P-450 structure, including
On the basis of experiments with rat liver an overall hydrophobic nature (36% of the amino
peroxisomes, it has been suggested that there is an w acids in this protein have either aromatic or
hydroxylase in peroxisomes active toward Cz;-steroids hydrophobic sideshains) and a conserved cystein
(11).Attempts to confirm this in our laboratory and in residue (located at position 444 in the 27-hydroxylase)
the laboratory of Pedersen have failed, however. that is thought to be a ligand for the heme iron. The

Downloaded from www.jlr.org by guest, on May 14, 2019


In view of the lesser importance of the 2t% structure of the enzyme displays a similarity with the
hydroxylase in mammals, only the 27-hydroxylase will mitochondrial cholesterol sidechain cleavage enzyme
be discussed in this review. Early work by Bjdrkhem (34%) and to the adrenal mitochondrial 1lphydroxyl-
and Gustafsson (12, 13) and by Taniguchi, Hoshita, ase (33%).A signal sequence of 36 residues was found
and Okuda (14) and Okuda, Weber, and Ullrich (15) to precede the coding region and this signal sequence
established that this enzyme is a mixed function is common to sequences that direct proteins into the
oxidase containing cytochrome P-450. The enzyme is mitochondrion. The 27-hydroxylase was regarded as a
bound to the inner mitochondrial membrane. Thus member of a novel cytochrome P-450 gene family.
the activity was low with intact mitochondria and Several research groups in the bile acid field have now
NADPH as a co-factor (13, 14). Under such conditions decided to call the gene CW 27.
citric acid and isocitric acid, which are able to Andersson et al. (10) were also able to express the
penetrate the inner mitochondrial membranes, stimu- 27-hydroxylase activity in cultured monkey COS cells.
late 27-hydroxylation much more efficiently than 27-Hydroxylase enzyme activity in COSM6 cells trans-
NADPH (13, 14). It is evident that citric acid and fected with the cDNA was enhanced by co-transfection
isocitric acid generate NADPH inside the mitochon- of a plasmid encoding bovine adrenodoxin but not by
drial membranes. With leaking mitochondria, NADPH a plasmid encoding NADPH cytochrome P-450 reduc-
stimulates the reaction as efficiently as isocitrate. tase. As could have been expected from previous work,
There is some evidence for the presence of different mRNA measurements showed that the enzyme was ex-
transport mechanisms for different substrates through pressed in several different organs and tissues (10).
the mitochondrial membranes (16). This may be of Most interesting, the abundance of the mRNA for the
some importance for the different rates of hydroxyla- enzyme paralleled the cholesterol biosynthesis capacity
tion of different substrates by intact liver of the tissues that were assayed. Thus the mRNA levels
mitochondria. were high in the liver, duodenum, adrenal gland, and
Pedersen, Oftebro, and Vinngird (17) and Sato et lung and less abundant in the kidney and spleen.
al. (18) reported simultaneously that small amounts of The liver mitochondrial 27-hydroxylase seems to be
cytochrome P-450 could be solubilized from the inner of little importance for the regulation of bile acid
membranes of rat liver mitochondria. The cytochrome biosynthesis and the composition of bile acids formed.
P-450 fraction obtained was active toward cholesterol The activity is high compared to that of the enzyme
as well as toward 5@holestane-W,7a, 12a-triol in the that catalyzes the rate-limiting step, cholesterol 7a-
presence of ferredoxin, ferredoxin reductase, and hydroxylase. It has been reported that the rat enzyme
NADPH. Ferredoxin and ferredoxin reductase were is slightly inhibited by biliary drainage and that it is
active regardless of whether they were isolated from not affected by, for example, starvation and phenobar-
rat liver mitochondria or bovine adrenal mitochondria bital treatment (12). Biliary obstruction seems to have
(19). Pedersen, Bjdrkhem, and Gustafsson (19) different effects on the rat mitochondrial 27-hydroxyl-

458 Journal of Lipid Research Volume 33, 1992


ase, depending on the substrate (25). Whether these considerably more polar than cholesterol. Direct
effects are due to effects on the 27-hydroxylase per se evidence for this hypothesis is still lacking, however.
or to effects on the transfer of steroids to the site of Pedersen, Holmberg, and Bjdrkhem (32) and
the enzyme could not be established. Saarem and Bjorkhem et al. (33) showed that a crude preparation
Pedersen (26) was well as Dahlbick (27) reported that of cytochrome P-450 from rat liver mitochondria was
the level of the enzyme is higher in female than in able to catalyze not only 27-hydroxylation of various
male animals. Cholic acid and starvation was found to C2rsteroids but also 25hydroxylation of cholesterol
reduce the amount of mitochondrial cytochrome P- and vitamin Ds. A kinetic study suggested that dif-
450-27 and the catalytic activity by 30-60%. There were ferent enzymes, or at least different binding sites, may
no parallel changes in the mRNA encoding the en- be involved in the 25- and 27-hydroxylations (33). In
zyme however (27). accordance with this contention, Dahlbick (34)
The rat, human, and pig 27-hydroxylases have showed that monoclonar antibodies against a purified
catalytical properties similar to those of the rabbit en- 27-hydroxylase inhibited 27-hydroxylation but not 25-
zyme (22-24). The deduced amino acid sequences of hydroxylation by a reconstituted system containing
the rat and the human enzymes indicated that it is highly purified, apparently homogeneous mitochon-
73% and 8l%, respectively, identical to the rabbit en- drial cytochrome P-450, ferredoxin, and ferredoxin
zyme (23, 28). It may be mentioned in this connection reductase. In contrast to these results, however,
that Raza and Avadhani (29) isolated two bnaph- Ohyama et al. (35) reported that partial denaturation
thoflavone-inducible species of cytochrome P-450 from by heating and treatment of the enzyme by N-
rat liver that possessed 27-hydroxylase activity in addi- bromosuccinimide inactivated the two enzyme ac-
tion to hydroxylase activity toward various xenobiotics. tivities in a similar manner. It has also been shown that

Downloaded from www.jlr.org by guest, on May 14, 2019


In contrast to the cytochrome P-450 fractions COS-cells transfected with the 27-hydroxylase plasmid
described above, the preparation studied by Raza and expressed both 27- and 25-hydroxylase activity (27). At
Avadhani (29) could be reconstituted with both least in these cells, the same gene must therefore be
microsomal NADPH-cytochrome P-450 reductase and responsible for both activities.
mitochondrial ferredoxin + ferredoxin reductase. Post-translational changes in the primary gene
The possibility has been discussed that the 27- product or presence of a specific 27-hydroxylase in ad-
hydroxylase may be of importance for the overall dition to the more nonspecific 27-hydroxylase (posses-
regulation of cholesterol biosynthesis (for a review, see sing both 27- and 25-hydroxylase activity) may explain
ref. 30). One of the products, 27-hydroxycholesterol,is the apparently conflicting results from the different
thus known to be a potent inhibitor of the rate-limit- groups. Another possibility is that the binding of non-
ing enzyme in cholesterol synthesis, the HMGCoA polar substrates like cholesterol and vitamin D3 to the
reductase (31). The finding by Andersson et al. (10) active site of the enzyme may differ from the cor-
that the abundance of the mRNA for the enzyme responding binding of more polar substrates like 5 k
paralleled the cholesterol biosynthesis capacity of the cholestane-3a,7a,12a-triol.In accordance with this
tissues is also in agreement with this possibility. Since it latter hypothesis we have found that cyclosporin in-
is possible to survive a complete or almost complete hibits 27-hydroxylation (and 25-hydroxylation) of cho-
lack of the enzyme activity for a life-time (cf below), it lesterol and 25-hydroxylation of vitamin D but not
is evident that the enzyme activity cannot be obligatory 27-hydroxylation of more polar Czrsteroids catalyzed
for the down-regulation of cholesterol synthesis in by an apparently homogeneous cytochrome P-450
various tissues. In recent work from this laboratory, the preparation from rabbit liver (H. Dahlbick, I.
possibility was excluded that a mitochondrial 27- Bjorkhem, and H. Princen, unpublished observation).
hydroxylation is of importance in the down-regulation
of HMGCoA reductase by dietary cholesterol in mice Conversion of 27-hydroxysteroidto 27carboxysteroid
(E. Lund and I. Bjorkhem, unpublished results). It was The liver has a very high capacity to catalyze oxida-
shown that 27-hydroxylation of 26,26,26,27,27,27-?H~- tion of 27-hydroxysteroids into the corresponding 27-
cholesterol in mouse liver mitochondria was associated carboxysteroids. Three different systems have been
with a marked isotope effect. When such deuterated described.
cholesterol was added to the diet, it was able to sup- In 1965 Okuda and Danielsson (36) synthesized the
press HMGCoA reductase as efficiently as unlabeled 27-aldehyde, believed to be the intermediate in the
cholesterol. above conversion. This aldehyde was efficiently oxidiz-
It may be speculated that the 27-hydroxylase may be ed into 3a,7a,l2a-trihydroxy-5~-cholestanoic acid (tri-
involved in the transport of cholesterol out from the hydroxycoprostanoic acid, (THCA) in the cytosolic,
cell. Thus 27-hydroxycholesterol and, in particular, its microsomal, and mitochondrial fractions of a rat liver
oxidation product 3khydroxy-5cholestenoic acid are homogenate. The soluble. and the mitochondrial frac-

Bjiirlthem Mechanism of degradation of steroid side chain in formation of bile acids 459
tions were most efficient. The soluble 5bholestane- fraction together with the cytosol and ATP (46). The
3a,7~~,12~~,27-tetrol-NADdehydrogenase and 3a,7a, final oxidation and thiolytic cleavage were reported to
12a-trihydroxy-5~cholestan-27-al-NAD-dehydrogenase be catalyzed by the mitochondrial fraction (45, 47, 48)
were later purified by Okuda and Takigawa (37-39). It but also by the soluble fraction (45, 47) and by the
was suggested that the enzymes could be identical to al- microsomal fraction combined with the 100,000 g su-
cohol and aldehyde dehydrogenase, respectively. In ac- pernatant (48). In these early experiments no attempts
cordance with this, Waller, Theorell, and Sjovall (40) were made to separate out other subcellular fractions
and Bjorkhem, Jornwall, and Akesson (41) reported and marker enzymes were not used to check for cross-
that recrystallized alcohol dehydrogenase from horse contamination of the fractions.
liver was able to catalyze oxidation of the tetrol into the In view of the controversial results, the subcellular
carboxylic acid. The SS isoenzyme was found to be most localization of the enzyme system responsible for the
efficient (41). overall conversion of THCA into cholic acid and of
In accordance with the investigation by Okuda and DHCA (3a,7adihydroxy-5bholestanoic acid, dihy-
Danielsson (36), Dahlback et al. (42) reported that droxycoprostanoic acid) into chenodeoxycholic acid
rabbit liver mitochondria contain an NADdependent was reinvestigated by Pedersen and Gustafsson (49)
enzyme system capable of catalyzing oxidation of the and by Pedersen's group in collaboration with the
tetrol into the carboxylic acid. author (50, 51). The initial experiments showed that a
Anderson et al. (10) and Dahlback and Holmberg peroxisome-enriched fraction from rat liver had the
(43) showed that the purified mitochondrial cyto- highest capacity to catalyze conversion of THCA into
chrome P-450-27 was able to catalyze formation of cholic acid. When THCA and DHCA were incubated
3a,7a,l2a-trihydroxy-5~holestanoicacid from 5p- with different subcellular fractions of rat liver, the

Downloaded from www.jlr.org by guest, on May 14, 2019


cholestane-3a,7a,12al27-tetro1when reconstituted highest specific activities for conversion into cholic
with ferredoxin, ferredoxin reductase, and NADPH. acid and chenodeoxycholic acid were observed in a
The possibility was clearly excluded that the enzymatic light mitochondrial fraction that also contained the
activity was derived from contaminating mitochondrial highest activity of the peroxisomal marker enzymes
and cytosolic dehydrogenases. The intermediate al- (50, 51). After separation of the light mitochondrial
dehyde could not be isolated. It is possible, however, fraction on sucrose or Nycodenz gradients, it was
that an intermediate aldehyde may be dismutated to shown that the fractions containing the highest
give equal amounts of the tetrol and the carboxylic peroxisomal marker activities also catalyzed formation
acid. of cholic acid from THCA and chenodeoxycholic acid
The relative importance of the three systems is not from DHCA. The conversion required the presence of
known. Sjdvall, Anderson, and Lieber (44) showed NAD, CoA, ATP and Mg". With peroxisomes prepared
that deer mice, which genetically lack soluble alcohol on sucrose gradient, FAD stimulated the reaction. The
dehydrogenase, have the same bile acid pool size and reaction was stimulated by KCN and unaffected by in-
composition as animals with normal levels of the en- hibitors of the mitochondrial respiratory chain. Small
zyme. It was therefore concluded that the soluble al- amounts of 24OH-THCA could be isolated from in-
cohol dehydrogenase cannot be obligatory for bile cubations with THCA.
acid biosynthesis. The findings were consistent with the view that the
Inasmuch as the 27-hydroxylation step occurs inside reaction sequence for oxidative cleavage of the C y 7
the mitochondria, it is attractive to suggest that one or steroid side-chain is similar to that of the peroxisomal
both of the two mitochondrial systems are most impor- boxidation of fatty acids, involving a THCA-CoA syn-
tant for the conversion. Because the carboxylic acid is thetase, an FADdependent oxidase, a hydratase, an
considerably more polar than its precursor, it is pos- NADdependent dehydrogenase, and a thiolase (Fig.
sible that the conversion is of importance for the trans- 2). A AZ4-unsaturated steroid should be an inter-
port of the bile acid intermediate from the mediate in this reaction. Indirect evidence for the in-
mitochondria. termediate formation of such a compound was
obtained from experiments performed in the presence
Peroxisomal Boxidation of coprostanoic acids of 'H20 or in an atmosphere of "Op (52). In the
Early experiments by Masui and Staple (45) showed former case there was an incorporation of one atom of
that 3a,7a,12a,24tetrahydroxy-5~holestanoic acid is deuterium into the 25-position, whereas in the latter
a probable intermediate in the conversion of THCA case there was no incorporation of isotope. Using
into cholic acid and that the 24hydroxylation step was specific incubation conditions without NAD, it was
catalyzed by the mitochondrial fraction of a rat liver later possible to show the direct conversion of THCA
homogenate. It was subsequently shown that this into Az4-THCA (53). It was also shown that the AZ4-
hydroxylation was also catalyzed by the microsomal double bond had the ZZ-configuration.

460 Journal of Lipid Research Volume 33, 1992


The first step in the overall conversion of THCA After purification, the reduced denatured enzyme
into cholic acid is the activation to yield a CoA deriva- migrated as a single 50 kDa protein band by sodium
tive, The microsomal fraction of a rat liver homoge- dodecyl sulfate-polyacrylamide gel electrophoresis. A
nate was found to have the highest catalytic activity, similar molecular mass was obtained for the native en-
whereas the peroxisomal fraction had little or no zyme by HPLC gel filtration. The purified enzyme was
capacity to catalyze this conversion (54). The THCA- found to use glycine, taurine, and 2-fluoro-&alanine
CoA oxidase was found to be a specific system, not but not alanine as substrates. It was suggested that a
identical to that involved in peroxisomal fatty acid single monomeric enzyme is responsible for conjuga-
oxidation. In contrast to the fatty acid oxidation sys- tion of bile acids with both glycine and taurine in
tem, clofibrate did not stimulate the THCA oxidase human liver.
(55) and treatment of rats with partially hydrogenated
marine oil was found to have different effects on the THE 25-HYDROXYLASE PATHWAY
two systems (56). Schepers et al. (57) described a par-
tial purification of the THCA-CoA oxidase from rat In addition to the 27-hydroxylase pathway, there is
liver peroxisomes and showed that it was different an alternative mechanism for degradation of the
from palmitoyl-CoA oxidase. The THCA-CoA oxidase steroid sidechain in 5bholestane-3a,7a,lPa-triol. It
had an apparent molecular mass of 139 kDa and con- was shown early on in both rat (64) and human (4)
sisted mainly, if not exclusively, of one polypeptide that 5bholestane-3a,7a, 12a-triol is efficiently 25-
component of 69 kDa. hydroxylated in the microsomal fraction. Shefer et al.
The next two steps in the conversion are catalyzed (63) and Salen et al. (64) further demonstrated that
by a bifunctional enzyme, enoyl-CoA hydratase/ khy- the product, 5~cholestane-3a,7a,12a,25-tetrol, is

Downloaded from www.jlr.org by guest, on May 14, 2019


droxyacyl-CoA dehydrogenase. This enzyme seems to converted into 5(%cholestane-3a,7a,l2a,24a,25-pentol,
be the same as that involved in peroxisomal fatty acid 5bholestane-3a,7a,12a24P,25-pentol, 5bholestane-
oxidation. Using photoaffinity labeling of isolated intact 3a,7a,12a,23,25pentol, and 5~cholestane-3a,7a, 1213,
rat liver peroxisomes with tritium-labeled 7,7-azo- 25,26-pentol in the presence of microsomes fortified
3a,12a-dihydroxy-5Pcholestan-27-oyl-CoA, Gengenbac- with NADPH. In the presence of NAD, 5bholestane-
her et al. (58) showed an incorporation of radioactivity 3a,7a,12a,24P,25-pentol, but not the other 5Pcholes-
into polypeptides that immunoprecipitated with an- tanepentols formed, is converted into cholic acid by
tibodies toward both the bifunctional enzyme and the soluble enzymes (Fig. 3). The latter conversion must
thiolase involved in peroxisomal Poxidation of fatty be assumed to involve acetone. These experiments
acids. demonstrated the existence of a new pathway for side-
The final step in the sequence of reactions leading chain degradation in cholic acid biosynthesis that does
to a conjugated bile acid is catalyzed by a bile acid- not involve hydroxylation at C-26 or the participation
CoA:amino acid N-acyltransferase. Kase et al. (59) and of mitochondria. The relative importance of this path-
Kase and Bjorkhem (60) showed that the highest way has been a matter of controversy (cf below). Since
specific amidation activity of both choloyl-CoA and
chenodeoxycholoyl-CoA was always found in the most
peroxisome-rich fractions of a rat liver homogenate.
The microsomal fraction contained less activity, while
the cytosol was inactive. In previous work substantial
amounts of enzyme activity were found in the cytosol.
This may be due to leakage of peroxisomal enzymes
HO' c$-
,c$
l -OH-

into the cytosol during the preparation of the different I I1


subcellular fractions. Striking differences were o b
served in the & values and the saturation concentra-
tions for glycine and taurine in the peroxisomal
system. It was suggested that most of the de novo syn-
thesized bile acids conjugated to taurine by the
peroxisomal systems, whereas the bile acids decon-
jugated in the gut and recirculation to the liver might
be activated and amidated by the microsomal enzyme
I11 IV
system prior to biliary secretion.
Very recently Johnson et al. (61) isolated a bile acid- Fig. 3. Formation of cholic acid from 5~holestane-3a,7a,l2a-triol
by the 25-hydroxylase pathway described by Salen et al. (64) I = 5p-
CoA:amino acid N-acyltransferase from the soluble cholestane-3a,7a,l2a-triol; I1 = 5~holestane-Sa,7a,12a,25-tetrol;
fraction of homogenized purified frozen human liver. 111 = 5pcholestane-3a,7a,12a,24(S),25-pentol;IV = cholic acid.

Bjiirlzhem Mechanism of degradation of steroid side chain in formation of bile acids 461
there is little or no 25-hydroxylation of 5bholestane-
3a,7a-diol in liver microsomes, the 25-hydroxylase
pathway cannot be of importance in the formation of
chenodeoxycholic acid.
It may be mentioned that the microsomal 25-
HO O Y
hydroxylase is not active towards cholesterol (4). As
mentioned above, the mitochondrial fraction of a liver
homogenate is able to catalyze 25-hydroxylation of
27-hydroxylase

Pathwa/

HO014C CH,?4CH,
~
\ 25-hydroxylase
Pathway
C O / c14
H3

W H 3
cholesterol (13). The rate of this hydroxylation is, trapped a s
however, very low and it seems unlikely that a pathway 2.4-dinitrophenylhydrazone

involving 25-hydroxycholesterol is of importance in 14


CO2
the biosynthesis of bile acids. trapped with
phenethylamine

Fig. 4. Experimental approach used by Duane et al. (65, 66) to


RELATIVE IMPORTANCE OF THE 2 5 AND evaluate the relative importance of the 25- and the 27-hydroxylase
27-HYDROXYLASE PATHWAYS pathways in the biosynthesis of bile acids in rat and humans.

On the basis of the efficient 25-hydroxylation of 5k


cholestane-3a,7a,l2a-triolin human liver microsomes
and the accumulation of 5fkholestane-3a,7a,l2a,25-
side-chain. Thus 5~holestane-3a,7a-diolis the sub-
tetrol in bile and feces of patients with cerebroten-
strate for the 27-hydroxylase in the formation of
dinous xanthomatosis (CTX, see below), Salen et al.

Downloaded from www.jlr.org by guest, on May 14, 2019


chenodeoxycholic acid and 5bholestane-3a,7a,12a-
suggested that the 25-hydroxylase pathway may be the
triol is the corresponding substrate in the formation of
major mechanism for biosynthesis of cholic acid in
cholic acid. This pathway is most probably
humans (64). There are, however, a number of experi-
predominant in rats but the situation may be different
mental findings that suggest that the 27-hydroxylase
in humans.
pathway is most important in the biosynthesis of both
Since the mitochondrial 27-hydroxylase has a broad
cholic acid and chenodeoxycholic acid.
substrate specificity (4, 12, 13), it is evident that path-
First, there are two known inborn errors of metabo-
ways may exist where the 27-hydroxyl group is intro-
lism in the 27-hydroxylase pathway in humans (cf
duced at a stage where only part of the nuclear
below). In both cases the metabolic block leads to ac-
changes have occurred. In the extreme case cholester-
cumulation of intermediates (or their metabolites) in
ol may be 27-hydroxylated prior to the introduction of
the 27-hydroxylase pathway. In one of the diseases
the 7a-hydroxyl group. Mitropoulos et al. (67) and
(CTX), one of the accumulated products is also an in-
Mitropoulos and Myant (68) showed that there was a
termediate in the 25-hydroxylase pathway. Taken
small conversion of cholesterol into 27-hydroxy-
together, however, the metabolic consequences of the
cholesterol, 3@hydroxy-5cholenoic acid, lithocholic
metabolic blocks clearly support the contention that
acid, and chenodeoxycholic acid in rat liver mito-
the 27-hydroxylase pathway is the more important.
chondria fortified with cytosol (Fig.5 ) . Lithocholic
Second, the 25-hydroxylase pathway generates
acid is, however, a less efficient precursor to cheno-
acetone as a cleavage product, whereas the 27-
deoxycholic acid and all available evidence suggests
hydroxylase pathway generates propionic acid. In con-
that a pathway involving lithocholic acid as inter-
trast to acetone, propionic acid is rapidly oxidized to
mediate is of little or no importance under normal
carbon dioxide. By studying the relative formation of
conditions. Such a pathway may, however, be of some
radioactive carbon dioxide and acetone from choles-
importance under cholestatic conditions, for example.
terol labeled with 14C in the terminal methyl groups,
Thus it has been shown that urine from infants with
Duane et al. (65, 66) showed in both rat and human
biliary atresia and patients with liver disease contains
that the 25-hydroxylase pathway is a minor one under
elevated concentrations of 3fbhydroxy-5-cholenoicacid
normal conditions (Fig.4).
(for a review, see ref. 1).
Among the many possible alternative pathways in
SPECIES DIFFERENCES AND ALTERNATIVE S U E which the 27-hydroxyl group is introduced at a stage
STRATES FOR THE 27-HYDROXYLASE PATHWAY prior to completion of the nuclear changes (for a
review see ref. l ) , there is at present experimental sup-
In the sequence of reactions shown in Fig. 1, all the port for the presence of three specific such pathways
nuclear changes precede the changes in the steroid in human liver.

462 Joumal of Lipid Research Volume 33, 1992


Most of the intermediates from this compound to

HO&- cholic acid were identified in the incubation medium


of these cells and the amounts of the products were
shown to change in a characteristic manner by an al-
teration of conditions. According to these authors
(72), cholic acid would be formed from 7a,12a-
dihydroxy4cholesten-hne by the sequence of 27-
hydroxylation, oxidation, and degradation of the side-
chain and A-ring reduction. An alternative pathway to
cholic acid was also described including reduction of
the intermediate 7a,12adihydroxy-3-ox&holesten-
oic acid to form 3a,7a,12a-trihydroxy-5~holestanoic
acid (THCA) prior to side chain cleavage (72).
HO’
Whether or not bile acids are formed by the same
mechanism in normal hepatocytes as in the above
Fig. 5. Mitochondrial conversion of cholesterol into chenodeoxy-
cholic acid (67, 68).
hepatoblastoma cells is not known with certainty. The
fact that two of the described intermediates,
7a,l2a,27-trihydroxy4cholesten-hneand 7a,12adi-
hydroxy-&x&holestenoic acid, are present in
Pathway involving 7a,27dihydroxy4cholesten-3-one human blood supports the contention that the path-
as intermediate way is significant (73).

Downloaded from www.jlr.org by guest, on May 14, 2019


7a-Hydroxy4cholesten-3-one is an efficient s u b
strate for the mitochondrial 27-hydroxylase in both rat
Pathway involving 27-hydroxycholesterolas
and human liver (4, 11). The product of the mito-
intermediate
chondrial 27-hydroxylase, 7a,27dihydroxy-4-cholesten-
&ne’, is rapidly converted into both cholic acid and Human liver mitochondria are able to 27-hydroxy-
chenodeoxycholic acid in vivo in humans (69, 70). late cholesterol (4). Krisans et al. (74) have shown that
Good evidence that this pathway is of importance in 27-hydroxycholesterol can be further metabolized into
humans is our finding that the lack of the mito- 3~hydroxy-5-cholenicacid in rat liver peroxisomes.
chondrial 27-hydroxylase in patients with CTX (see The cofactor requirement was the same as that in the
below) leads to a substantial accumulation of 7a- peroxisomal conversion of THCA into cholic acid
hydroxy4cholesten-3ne and its metabolite, 7a,12a- (74). Anderson, Kok, and Javitt (75) reported that 27-
dihydroxy4-cholesten-&ne (71). It is not known with hydroxycholesterol is rapidly converted into both
certainty how 7a,27dihydroxy4cholesten-3-oneis fur- cholic and chenodeoxycholic acid in vivo in humans.
ther metabolized. One alternative would be comple- 27-Hydroxycholesterol and 3~hydroxy-5-cholenoic
tion of the sidechain cleavage prior to further nuclearacid intermediates in the above pathway are present in
changes. Axelsson, Mbrk, and Everson (72) recently relatively high concentrations in human blood (76-
described such a pathway in cultured HepG2 cells. 78). We have found that the concentration of 27-
They isolated and identified all intermediates from 7a-hydroxycholesterol is considerably lower in the
hydroxy-4-cholesten-3-one to chenodeoxycholic acid circulation of rats and rabbits than in humans (I.
from the incubation medium. In this pathway side- Bjorkhem, unpublished observation) possibly indicat-
chain degradation was completed prior to 5breduc- ing that pathways involving 27-hydroxycholesterol are
tion of the A-ring. less important in the former species.
Axelsson and Sjbvall (73) and Axelsson, Mijrk, and
Sjbvall (76) have denoted the pathway to chenode-
Pathway involving 7a,12a,27-trihydroxyP oxycholic acid starting with 27-hydroxylation of choles-
cholesten-3-one as intermediate terol and proceeding via Csracids the “acid” pathway.
The accumulation of 7a,l2a-dihydroxy4-cholesten- The one starting with 7a-hydroxylation of cholesterol
&ne in patients with a lack of the mitochondrial 27- was called the “neutral” pathway. Since they found that
hydroxylase indicates that this compound may also be the levels of the circulating intermediates in the
an important substrate for the 27-hydroxylase in vivo neutral pathway were increased after cholestyramine
(71). In accordance with this, a pathway starting with treatment, whereas the intermediates in the acid path-
sidechain oxidation of 7a,12a-dihydroxy-4-cholesten- way were unaffected, they predicted that the two path-
&ne was very recently described in HepG2 cells (72). ways were regulated separately. They also studied the

Bjkkhem Mechanism of degradation of steroid side chain m formation of bile acids 463
behavior of 7a-hydroxy-3-ox&holestenoic acid in INBORN ERRORS IN SIDE-CHAIN CLEAVAGE
the human circulation under different conditions
(73). This compound may be formed in both the There are two known types of inborn metabolic er-
“neutral” and the “acid” pathway. In accordance with rors that affect cholesterol sidechain cleavage,
this, the level of this acid was increased in the circula- cerebrotendinous xanthomatosis (CTX) and various
tion after cholestyramine treatment but not to the peroxisomal disorders. In CTX there is a specific
same degree as 7a-hydroxy-4cholesten-3-one (73). defect in one of the enzymes. In the peroxisomal dis-
Further support for the presence of a pathway start- orders there is a lack of intact peroxisomes and thus
ing with 27-hydroxylation comes from work by Princen also a lack of the peroxisomal enzymes necessary for p
et a1 (79) and from collaborative work of H. Dahlback, oxidation of cholestanoic acids.
1. Bjorkhem, and H. Princen (unpublished observa-
tion). It was found that cyclosporin selectively inhibits Cerebrotendinousxanthomatosis
27-hydroxylation of cholesterol but not 27-hydroxyla- This rare, inherited lipid storage disease is charac-
tion of other 7a-hydroxylated bile acid intermediates terized by xanthomas, progressive neurological dys-
in liver mitochondria and in a reconstituted purified function, cataracts, and the development of
cytochrome P450 system. Cholesterol 7a-hydroxylase xanthomatous lesions in the brain and lung (for a
was not affected by the inhibitor. Hepatocytes from rat general review, see ref. 81). In contrast to other dis-
and human hepatoblastoma cells (HepG2 cells) eases with tendon xanthomatosis, plasma cholesterol
responded with decreased bile acid formation when levels are remarkably low. Large deposits of cholesterol
exposed to cyclosporin. Such a decrease would be ex- and cholestanol are present in most tissues. In 1974,
pected if part of the bile acids is formed in a pathway Setoguchi et al. (82) made the key discovery that CTX

Downloaded from www.jlr.org by guest, on May 14, 2019


involving 27-hydroxylation as a first and rate-limiting patients have a defect in bile acid biosynthesis, with in-
step. It may be mentioned that cyclosporin has a hy- complete oxidation of the C27-steroid sidechain, lead-
percholesterolemic effect and it may be speculated ing to excretion of large amounts of Cn~bilealcohols
that this may be due in part to a blocking of the “acid” in bile, feces, and urine. The formation of bile acids,
pathway for bile acid biosynthesis. in particular chenodeoxycholic acid, was reduced.
If 27-hydroxycholesterol is an intermediate in a There has been a controversy in the past between
pathway to cholic and chenodeoxycholic acid, the 7a- our laboratory and that of Salen et al. concerning the
hydroxyl group may be introduced at different stages. location of this defect (for a general review, see ref.
It was recently reported (80) that 27-hydroxy- 81). It is however, now established that the basic meta-
cholesterol is efficiently 7a-hydroxylated in pig liver bolic defect is located to the mitochondrial 27-
mitochondria. In preliminary experiments we found hydroxylase. We have shown, for example, that
little or no 7a-hydroxylase activity toward 27-hydroxy- fibroblasts from CTX patients do not express this en-
cholesterol in human liver mitochondria (I. Bjorkhem, zyme activity and that fibroblasts from heterozygotes
E. Reihnkr, and K. Einarsson, unpublished observa- have an activity about 50% of normal (7). Very recent-
tions). Significant such activity was found in the ly Cali et al. (83) showed that the 27-hydroxylase gene
microsomal fraction of a homogenate from human in patients with CTX contained two cys-arg muta-
liver. Another group (J. Shoda, A. Toll, M. Axelsson, F. tions. When genes with these mutations were ex-
Pieper, K. Wikvall, and J. Sjovall, unpublished observa- pressed in COScells, the expressed enzyme was found
tions) found significant 7a-hydroxylase activity in both to be inactive.
the microsomal and mitochondrial fractions. Whether The lack of the mitochondrial 27-hydroxylase leads
or not the microsomal 7a-hydroxylase active towards to extensive accumulation of a number of substrates
27-hydroxycholesterol is the same as that active for the enzyme, such as 7a-hydroxy-4-cholesten-3-one
towards cholesterol is not known. and 5~cholestane-3a,7a,12a-triol. The accumulation
The relative importance of the pathway including of 7c~-hydroxy-4-cholesten-30nemay be due in part to
27-hydroxycholestero1 as an intermediate is not known the up-regulated cholesterol 7a-hydroxylase (see
and one can only speculate about this. According to below) which leads to increased levels of both 7a-
the work by Axelson and Sjovall (73) and Princen et hydroxycholesterol and 7a-hydroxy-4-cholesten-3-one
al. (79), this pathway may be responsible for up to in the liver and the circulation (81, 84, 85). The u p
50% of all bile acids formed in human liver under regulation of the cholesterol 7a-hydroxylase is due to
basal conditions. In a situation where bile acid biosyn- the low formation of primary bile acids, in particular
thesis is up-regulated, e.g., by a bile fistula or by chenodeoxycholic acid. Among the primary bile acids,
cholestyramine treatment, the contribution of the chenodeoxycholic acid seems to be the most efficient
“acid” pathway may be considerably less (73). suppressor of the human cholesterol 7&hydroxylase

464 Journal of Lipid Research Volume 33, 1992


(86). Since bile acids are involved also in the regula- Also other metabolites of 5fkholestane-3a,7a, 1201-
tion of the HMG-CoA reductase (1, 87, 88), cholester- triol, such as 5fkholestane-3a,l2a,12~~,24a-25pentol
ol synthesis is also increased in CTX. As a and 5~holestane-3a,7a,l2a,23-25-pentol,are excret-
consequence, LDL turnover is increased (89). The ed. In total, gram amounts of all the above bile alco-
possibility has been discussed that the increased cho- hols are excreted daily in bile and feces. Treatment
lesterol synthesis and LDL-turnover in CTX may be in with chenodeoxycholic acid suppresses the upregu-
part a direct effect of the lack of 27-hydroxycholesterol lated cholesterol 7a-hydroxylase and results in a drasti-
or its metabolites (30, 73). Treatment of patients with cally reduced formation of bile alcohols and
chenodeoxycholic acid seems to normalize cholesterol cholestanol (90).
synthesis, however (90).
Because 7a-hydroxyPcholesten-%ne cannot be Peroxisomal disorders
metabolized by the usual pathway to bile acids, more The most serious of the peroxisomal diseases affect-
unusual pathways are used (Fig. 6 ) . We have shown ing bile acid synthesis is Zellweger’s disease (for a
that one product of 7a-hydroxy-kholesten-%ne is general review, see ref. 93). This rare congenital disor-
cholestanol (81). This compound is formed by der is characterized by multiple craniofacial abnor-
dehydration of 7a-hydroxy~holesten-3-oneto 46 malities, generalized hypotonia, central nervous system
cholestadien-%ne by a specific enzyme in liver abnormalities, hepatomegaly, and renal cortical cysts.
microsomes (91). The latter steroid is rapidly con- Most often the afflicted infants die within 6 months.
verted into cholestanol (92). It appears likely that at Other peroxisomal disorders also affecting bile acid
least a substantial part of the cholestanol accumulating biosynthesis are infantile Refsum’s disease, and neona-
over the decades in the brain and tendons of patients tal adrenoleukodystrophia (93). Patients with Zell-

Downloaded from www.jlr.org by guest, on May 14, 2019


with CTX is formed from accumulated bile acid inter- weger’s disease lack intact peroxisomes and most of
mediates. the peroxisomal enzymes are reduced or absent. Con-
Because the accumulated 5bholestane-3a,7a,12a- sequently these patients have several severe metabolic
triol cannot be metabolized by the usual pathway to disturbances (reduced capacity to oxidize fatty acids,
bile acids, the 25-hydroxylase pathway may be used prostaglandins, phytanic acid, reduced synthesis of
(Fig. 7). Since the enzymes involved in this pathway plasmalogens) (93).
appear to have a limited capacity, some of the inter- In accordance with our finding that peroxisomes
mediates in this pathway accumulate and are excreted contain the enzymes involved in bxidation of the
in bile and feces. Thus patients with CTX excrete high CoA derivative of trihydroxycoprostanoic acid, patients
amounts of 5~holestane-3a,7a,l2a,Z5-tetrol(81, 82). with Zellweger’s disease, infantile Refsum’s disease,

cho 1 e s t e r o 1 -&
HO
n ,

‘ ”OH
chenodeoxy
cholic acid

cholestanol

Fig. 6. Metabolic consequence of the accumulation of 7~-hydroxy4cholesten-3-onein patients with CTX.


Mechanism of formation of excess cholestanol (81).

Bjtirkhem Mechanism of degradation of steroid side chain in formation of bile acids 465
cholesterol -#-
- &e
HO HO'
cilohT:&

HO'
acid

HO'
&OH

"OH HO'
& 8 "OH
OH

HO' "OH
OH .....,......,.

Fig. 7. Metabolic consequence of the accumulation of 5~holestane-Sa,7a,l2a-uiolin patients with CTX.


Mechanism of formation of C ~ b i l ealcohols and utilization of the 25-hydroxylase pathway for formation of
cholic acid.

Downloaded from www.jlr.org by guest, on May 14, 2019


and neonatal adrenoleukodystrophia accumulate patients, different labeled intermediates in bile acid
THCA and a number of metabolites of this compound biosynthesis, carrying an intact Czrsteroid sidechain,
(for general reviews, see refs. 93 and 94). The most im- were found to be rapidly converted into THCA but
portant metabolites that accumulate in the above dis- then only very slowly to cholic acid (95, 96). A liver
eases are summarized in Fig. 8. One interesting and biopsy from a patient with Zellweger's disease had no
quantitatively important metabolite of THCA acid is a significant ability to convert THCA into cholic acid
dicarboxylic Cwacid (94). The mechanism behind the (96). There was, however, some conversion of 301,7a-
formation of this chain-elongated derivative is not dihydroxy-5f3-cholestanoicacid into chenodeoxycholic
known. It should be emphasized that the metabolic acid. The latter finding may explain why there is little
block is not complete since patients with above or no accumulation of 301,7~tdihydroxy-5~holes-
peroxisomal disorders also form some cholic acid. In tanoic acid in this disease. Another explanation is that
collaborative studies together with Pedersen and Kase 301,7~xdihydroxy-5~holestanoic acid is efficiently 1201-
we have shown, however, that the cholic acid and hydroxylated by the human liver (97). Yet, another ex-
chenodeoxycholic acid pool sizes are drastically planation was presented by Axelson et al. (72) who
reduced (95, 96). After administration to Zellweger found an accumulation of 7c~hydroxy-3-ox&holes

&COOH fl COOHCOOH

HO' "OH HO' "OH HO' "OH


OH
Fig. 8. Metabolic consequence of the accumulation of THCA in patients with the Zellweger disease. Only
the quantitatively most dominating metabolites of THCA are shown.

466 Journal of Jipid Research Volume 33, 1992


the products was identical to chenodeoxycholic acid
and only traces at the most could be identical to cholic
acid. The results suggested that healthy human sub-
jects, like other mammalian species studied, have little
or no capacity to convert sitosterol into the normal
Cpqbile acids.
Sitosterol Stigmasterol Campesterol
Several previous studies have shown that sitosterol is
Fig. 9. Structures of the most important phytosterols. converted into polar compounds in the bile acid frac-
tion of rat bile (100, 104, 105). Very recently we were
able to identify most of these products (106, 107). The
tenoic acid and its hydrolyzed product, 3-0x0-4,6- major part of the I4C radioactivity recovered as bile
cholestadienoic acid, in blood from a patient with this acids in bile after intravenous administration of [4
disease. This finding suggests that 7a-hydroxy-30x04 l4C1sitosterol to female Wistar rats was found to be
cholestenoic acid could be the substrate for the considerably more polar than cholic acid and only
peroxisomal sidechain degradation instead of 3a,'7a- trace amounts had chromatographic properties similar
dihydroxy-5@holestanoic acid. to those of cholic acid and chenodeoxycholic acid. It
In any case, it is evident that the accumulation of was shown that the polar products were di- and
THCA acid and its metabolites in the peroxisomal dis- trihydroxylated Czl-bile acids.
eases reflects the importance of the peroxisomes for Using mass spectrometry, NMR, stereospecific
the last step in the biosynthesis of cholic acid from dehydrogenases and reagents, the major trihydroxy-
cholesterol. lated Czl-bile acids were identified as 5ppregnan-

Downloaded from www.jlr.org by guest, on May 14, 2019


3~,11f3,15~trio1-21-oic acid, 5ppregnan-Sa,l lp,15a-21-
oic acid, and 5Ppregnan-Sa,l lf3,16-triol-21-oic acid.
MECHANISM OF DEGRADATION OF THE The major dihydroxylated &-bile acid was identified
STEROID SIDE CHAIN OF PLANT STEROIDS by the same means as 5a-pregnan-3a,l2~l-diol-21-oic
acid (Fig. 10) (see refs. 97, 98).
The structure of the three most common plant Considerably less Czl-bile acids were formed from
sterols is shown in Fig. 9. These plant sterols are nor- labeled sitosterol in male than in female Wistar rats.
mal constituents of the human diet. They are ab- The Czl-bile acids formed in male rats did not contain
sorbed to a very limited extent from the intestine, a 15-hydroxyl group. Conversion of labeled sitosterol
circulate in plasma in low concentrations, and are ex- into Czl-bile acids also occurred in adrenalectomized
creted in bile (for a review, see ref. 81). and ovariectomized rats, indicating that endocrine tis-
In theory, the presence of an ethyl group at C-24 sues were not involved. Experiments with isolated per-
should prevent or at least obstruct conversion of the fused liver gave direct evidence that the overall
plant sterols to bile acids. The possibility must be con- conversion of sitosterol into C2l-bile acids occurs in
sidered that sitosterol is first dealkylated to cholesterol this organ. Attempts to demonstrate production of
and then converted into bile acids. Such a dealkylation Cnl-bile acids in isolated hepatocytes have failed
occurs in some worms and crabs (98, 99) but attempts hitherto.
to demonstrate such a pathway in mammals have After intravenous injection of deuterium-labeled
failed hitherto. In accordance with this, attempts to sitosterol in bile fistula female Wistar rats, the isolated
demonstrate conversion of sitosterol into normal C24- Czl-bile acids were found to contain very little isotope,
bile acids in rats (100) and monkeys (101) have failed.
In an early study, Salen, Ahrens, and Grundy (102)
reported an efficient formation of cholic and
chenodeoxycholic acid from intravenously admin-
istered [22,2S3H]sitosterol in humans. In a reinves-
no .Acoon no

tigation with [4I4C]sitosterol we could not find any


significant conversion into labeled (&-bile acids in two
healthy subjects (103). In order to bypass the rate-
limiting step, the metabolic fate of "H-labeled '7a-
hydroxysitosterol was also studied. In this case there AcaoH
was a significant conversion into acid products in bile.
Although part of the labeled products had the chro-
matographic properties of cholic and chenode-
no no OY-
Fig. 10. Structures of the major Czi-bile acids formed from
oxycholic acid, further analysis showed that none of cholesterol and plant sterols in female Wistar ratS (97, 98).

Bjiirkhem Mechanism of degradation of steroid side chain m formation of bile acids 467
indicating a high dilution with endogenous material. CONCLUDING REMARKS
In view of the small amounts of sitosterol in the diet, it
appeared likely that Czl-bile acids may also be formed Three different mechanisms are known for the
from a more abundant precursor, such as cholesterol. degradation of the cholesterol side chain.
It was subsequently shown, using a mixture of [4 The major mechanism, the “27-hydroxylase path-
‘‘C]cholesterol and [22-3H]cholesterol, that there was way,” involves a primary attack by a mitochondrial
an efficient conversion of the administered material cytochrome P-450 system followed by peroxisomal
into the above C21-bile acids. The conversion was oxidation. Metabolic blocks in this pathway lead to ac-
surprisingly high, up to about 25%. It is thus evident cumulation of intermediates, with severe metabolic
that a blocking group at C24 is not obligatory for consequences.
degradation of the steroid side-chain beyond the C24 The “25-hydroxylase pathway” involves a primary at-
stage. It was further shown that campesterol, carrying a tack by a microsomal 25hydroxylase. This pathway has
methyl group in 24position, was also converted into a low capacity and seems to be of importance only
the above C21-bile acid in female Wistar rats (106). when the 27-hydroxylase pathway is blocked.
The mechanism for formation of the Czl-bile acids In addition to the above systems, a complementary
can only be speculated about at the present stage of system has been evolved in at least one mammalian
knowledge. The most likely mechanism seems to be a species that is able to degrade both cholesterol and
primary attack by a mixed function oxidase at C21, fol- plant sterols into highly polar Cgl-bile acids. It is prob-
lowed by further oxidation to give a C2lcarboxylic able that this degradation is initiated by attack of a 21-
group. The latter compound may then be further hydroxylase. The relative importance of the
oxidized to give a C21-bile acid as final product (Fig. hypothetical “21-hydroxylase pathway” in different

Downloaded from www.jlr.org by guest, on May 14, 2019


11).At present, however, there is no direct evidence of mammalian species is not known. I
this hypothetical “21-hydroxylase pathway.” In prelimi-
nary experiments we synthesized labeled 21- The author is most grateful to the following colleagues for
hydroxycholesterol and administered it to female reading the manuscript, for consuuctive criticism, and for
Wistar rats. Most of the polar products obtained were information concerning unpublished studies: Dr. Magnus
not identical to the above Cpl-bile acids. It is thus likely Axelson, Dr. Kyuchiro Okuda, Dr. Jan I. Pedersen, Dr. Hans
that some metabolic step(s) in the steroid nucleus Princen, Dr. Jan Sjbvall, and Dr. Kjell Wikvall. The author’s
precedes the 21-hydroxylation. It is tempting to sug- own work, referred to in the review, has been supported by a
grant from the Swedish Medical Research Council.
gest that the sex-specific 15-hydroxylation may
facilitate the formation of C2l-bile acids. Manusnipt received 21 Noumber I991
The high capacity of the female rat liver to convert
cholesterol into compounds considerably more polar
than the usual bile acids may be of some regulatory REFERENCES
importance in the overall cholesterol balance. The
role of Czl-bile acids in other mammalian species is 1. BjBrkhem, I. 1985. Mechanism of bile acid biosynthesis
uncertain, however. In humans there seems to be little in mammalian liver (Review). In Comprehensive
or no formation of such bile acids (E. Lund, K. Muri- Biochemistry. Vol. 12. H. Danielsson and J. Sjbvall,
editors. Elsevier Publishing Co., Amsterdam. 231-278.
Boberg, and I. Bjorkhem, unpublished observation). 2. Popjak, G., J. Edmond, F. A. L. Anet, and N. R. Eaton,
Jr. 1977. Carbon-13 NMR studies on cholesterol syn-
thesized from 13C-mevalonates.J. Am. Chem. SOC. 9 9
931-935.
3. Gustafsson, J., and S. Sjbstedt. 1978. On the stereo-
specificity of microsomal “26”-hydroxylationin bile acid
biosynthesis. J. Biol. C h . 253: 199-201.
4. BjBrkhem, I., J. Gustafsson, G. Johansson, and B.
Persson. 1975. Biosynthesis of bile acids in man:
hydroxylation of the C27-steroid side chain. J. Clin. In-
vest. 55: 478-486.

---
0-ox
HOOC 5. Hanson, R. F., P. Szczepanik, and G. C. Williams. 1980.
Stereochemistry of the side chain oxidation of 5p
cholestaneSa,7a,lfa-triolin man. J. Biol Chem. 255:
1483-1485.
R
6. Bjbrkhem, I., H. Danielsson, and J. Gustafsson. 1973.
Fii. 11. Suggested mechanism for formation of Czi-bile acids from On the effect of thyroid hormone on 26hydroxylation
phytosterols and cholesterol in female Wistar rats (97, 98). of Cgpsteroids in rat liver: FEBS Lett. 31: 20-22.

468 Journal of Lipid Research Volume 33, 1992


7. Skrede, S., I. Bjcrkhem, E. A. Kvittingen, M. S. Buch- mitochondrial cytochrome P450 from pig kidney and
mann, S. 0. Lie, C. East, and S. Grundy. 1986. liver catalyzing 26hydroxylation of 25hydroxyvitamin
Demonstration of 26hydroxylation of Cq7-steroids in D3 and C2’1-steroids. Biochem.J. 276: 427-432.
human skin fibroblasts and deficiency of this activity in 25. Gustafsson, J. 1978. Effect of biliary obstruction on 2 6
CTX. J. Clin. Invest. 78: 729-735. hydroxylation of Cpsteroids in bile acid synthesis. J.
8. Pedersen, J. I., H. Oftebro, and I. Bjdrkhem. 1989. Lipid Res. 19: 237-243.
Reconstitution of Cp7-steroid 26hydroxylase activity 26. Saarem, K., and J-I. Pedersen. 1987. Sex differences in
from bovine brain mitochondria. Biochem. Znt. 1 8 615- the hydroxylation of cholecalciferol and of 5bholes-
622. tane-3a,7a,l2a-triol in rat liver. Biochem.J. 247: 73-78.
9. Postlind, H., and K. Wikvall. 1989. Evidence for the for- 27. Dahlbick, H. 1989. Thesis. Uppsala University.
mation of 26hydroxycholesterol by cytochrome P450 Lindbergs Grafiska H. B.
in pig kidney mitochondria. Biochem. Biophys. Res. Com- 28. Usui, E., M. Noshiro, and K. Okuda. 1990. Molecular
mun. 159: 1135-1140. cloning of cDNA for vitamin D3 25hydroxylase from rat
10. Anderson, S., D. L. Davis, H. Dahlbick, H. Jdrnvall, liver mitochondria. FEBS Lett. 262 135-138.
and D. W. Russell. 1989. Cloning, structure, and expres- 29. Raza, H., and N. G. Avadhani. 1988. Hepatic
sion of the mitochondrial cytochrome P-450 sterol 2 6 mitochondrial cytochrome P-450 system: purification
hydroxylase, a bile acid biosynthetic enzyme. J. Biol. and characterization of two distinct forms of mito-
Chem. 264 8222-8229. chondrial cytochrome P450 from knaphthoflavone-in-
11. Thompson, S. L., and S. K. Krisans. 1985. Evidence for duced rat liver. J. Biol. Chem. 263: 9533-9541.
peroxisomal hydroxylase activity in rat liver. Biochem. 30. Javitt, N. B. 1990. 26HydroxycholesteroI: synthesis,
Biophys. Res. Commun. 1 3 0 708-716. metabolism, and biologic activities. J. Lipid Res. 31:
12. Bjdrkhem, I., and J. Gustafsson. 1973. @Hydroxylation 1527-1533.
of steroid sidechain in biosynthesis of bile acids. Eur. J. 31. Esterman, A. L., H. Baum, N. B. Javitt, and G. J. Dar-
Biochem. 36: 201-212. lington. 1983. 26Hydroxycholestero1: regulation of
13. Bjdrkhem, I., and J. Gustafsson. 1974. Mitochondrial w hydroxymethylglutaryl-CoA reductase activity in

Downloaded from www.jlr.org by guest, on May 14, 2019


hydroxylation of the cholesterol side chain. J. Biol. Chinese hamster ovary cell culture. J Lipid Res. 2 4
Chem. 249: 2528-2535. 1304-1 309.
14. Taniguchi, S., N. Hoshita, and K. Okuda. 1973. En- 32. Pedersen, J. I., I. Holmberg, and I. Bjdrkhem. 1979.
zymatic characteristics of CO-sensitive 26hydroxylase Reconstitution of vitamin D3 25-hydroxylase activity
system for 5~holestane-3a,7a,12a-triolin rat liver with a cytochrome P450 preparation from rat liver
mitochondria and its intramitochondrial localization. mitochondria. FEBS Lett. 9 8 394-398.
Eur.J.’Biochem. 40: 607-617. 33. Bjdrkhem, I., I. Holmberg, H. Oftebro, and J. I. Peder-
15. Okuda, IC,P. Weber, and V. Ullrich. 1977. Photochemi- sen. 1980. Properties of a reconstituted vitamin D3 2 5
cal action spectrum of the CO-inhibited 5fiholestane hydroxylase from rat liver mitochondria. J. Biol. Chem.
3a,7a, 12a-triol 26hydroxylase system. Biochem. Biophys. 255: 52445249.
Res. Commun. 7 4 1071-1075. 34. Dahlbick, H. 1988. Characterization of the liver
16. Gustafsson, J. 1976. On the heterogeneity of the mitochondrial cytochrome P-450 catalyzing the 26-
mitochondrial Cg7-steroid 26hydroxylation system. J. hydroxylation of 5bholestane-3a,7a,l2a-triol.Biochem.
Lipid Res. 17: 366-372. Biophys. Res. C?mmun. 157: 30-36.
17. Pedersen, J., H. Oftebro, and T. Vinngird. 1977. Isola- 35. Ohyama, Y., 0. Masumoto, E. Usui, and K. Okuda.
tion from bovine liver mitochondria of a soluble fer- 1991. Multi-functional property of rat liver mitochon-
redoxin active in a reconstituted steroid hydroxylating drial cytochrome P450. J. Bdochem. (Tokychem (Tokyo)
system. Biochem. Biophys. Res. Commun. 76: 666-673. 109: 389-393.
18. Sato, R., Y. Atsuta, Y. Imai, S. Taniguchi, and K. Okuda. 36. Okuda, K., and H. Danielsson. 1965. Synthesis and me-
1977. Hepatic mitochondrial cytochrome P-450: isola- tabolism of 5~holestane-Sa,7c~,12a-trio1-26al. Acta
tion and functional characterization. R o c . Natl. Acad. Chem. Scand. 19: 2160-2165.
Sn’. USA 74: 5477-5481. 37. Okuda, K., and N. Takigawa. 1969. The dehydrogena-
19. Pedersen, J. I., I. Bjdrkhem, and J. Gustafsson. 1979. 2 6 tion of 5&cholestane-3a,7a,12a,26tetrolby rat liver.
Hydroxylation of C27-steroids by soluble liver mitochon- Biochlm. Biophys. Acta. 176: 873-879.
drial cytochrome P-450. J. Biol. Chem. 254 64646469. 38. Okuda, IC, and N. Takigawa. 1968. Separation of 5p-
20. Wikvall, K. 1984. Hydroxylations in biosynthesis of bile cholestane-3a,7a,l2a,26-tetrol oxidoreductase, ethanol
acids: isolation of a cytochrome P-450 from rabbit liver oxidoreductase, 5~holestane-3a,7a,12a-triol-26al oxi-
mitochondria catalyzing 26hydroxylation of C27- doreductase and acetaldehyde oxidoreductase from rat
steroids. J. Biol. Chem. 259: 3800-3804. liver. Biochem. Biophys. Res. Commun. 3 3 788-793.
21. DahlbLk, K., and K. Wikvall. 1988. 25Hydroxylation of 39. Okuda, K., and N. Takigawa. 1970. Rat liver 5bholes-
vitamin D3 by a cytochrome PA50 from rabbit liver tane-3a,7a, 12a,26tetrol dehydrogenase as a liver alco-
mitochondria. Biochem.J. 252 207-213. hol dehydrogenase. Biochim. Biophys. Acta. 222:
22. Okuda, IC, 0. Masumoto, and Y. Ohyama. 1988. 141-148.
Purification and characterization of 5pcholestane- 40. Waller, G., H. Theorell, and J. Sjdvall. 1965. Liver al-
31~,7a,lBa-triol27-hydroxylase from female rat liver cohol dehydrogenase as a 3j3-hydroxy-5f3-cholanic acid
mitoch0ndria.J Biol. Chem. 263: 18138-18142. dehydrogenase. Arch. Biochem. @i%hys. 111: 671-684.
23. Cali, J. J., and D. W. Russell. 1991. Characterization of 41. Bjdrkhem, I., H. JBrnwall, and A. Akesson. 1974. Oxida-
human sterol 27-hydroxylase: a mitochondrial tion of *hydroxylated fatty acids and steroids by S S
cytochrome P450 that catalyzes multiple oxidations in isoenzyme of liver alcohol dehydrogenase. Biochem.
bile acid biosynthesis.J. Biol. Chem. 266: 77747778. Biophys. Res. Commun. 57: 870-875.
24. Bergman, T., and H. Postlind. 1991. Characterization of 42. Dahlbick, H., H. Danielsson, M. Gustafsson, J. Sjdvall,

Bj&khem Mechanism of degradation of steroid side chain in formation of bile acids 469
and R Wikvall. 1988. Conversion of 5P-cholestanoic 59. h e , B. F., K. Prydz, I. Bjbrkhem, and J. I. Pedersen.
acid by rabbit liver mitochondria. Biochem. Biophys. Res. 1986. Conjugation of cholic acid with taurine and
Commun. 153: 267-274. glycine by rat liver peroxisomes. Biochem. Biophys. Res.
43. Dahlbkk, H., and 1. Holmberg. 1990. Oxidation of 5 p Commun. 138: 167-1 73.
cholestane-3a,7a,l2a-triol into 3a,7a,l2a-trihydroxy- 60. Kase, B. F., and I. Bjbrkhem. 1989. Peroxisomal bile
5bholestanoic acid by cytochrome P-45026 from rabbit acid-CoAamino acid N-acyltransferase in rat liver. J.
liver mitochondria. Biochem. Biophys. Res. Commun. 167: Biol. Chem. 264: 9220-9223.
- ...391-395. . .___._ 61. Johnson, M. R., S. Barnes, J. B. Kwakye, and R. B.
44. SjGvall, J., S. H. G. Anderson, and C. S. Lieber. 1987 Diasio. 1991. Purification and characterization of bile
Bile acids in deer mice lacking liver alcohol dehy- acid-CoA:amino acid N-acyltransferase from human
drogenase. Biochim. Biophys. Acta. 836: 8-1 3. liver. J. Biol. Chem. 266: 10227-10233.
45. Masui, T., and E. Staple. 1966. The formation of bile 62. Cronholm, T., and G. Johansson. 1970. Oxidation of
acids from cholesterol. J. Biol. Chem. 241: 3889-3893. 5pcholestane-3a,7a,l2~~triol by rat liver microsomes.
46. Gustafsson, J. 1975. Biosynthesis of cholic acid in rat Eur. J. Biochem. 16: 373-381.
liver. 24Hydroxylation of 3a,7a,l2a-trihydroxy-5f3-cho- 63. Shefer, S., F. W. Cheng, B. Dayal, S. Hauser, G. S. Tint,
lestanoic acid. J. Biol. Chem. 250: 8243-8247. G. Salen, and E. H. Mosbach. 1976. A 25-hydroxylase
47. Masui, T., and E. Staple. 1965. The formation of cholic pathway of cholic acid in man and rat. J. Clin. Invest. 57:
acid from 3a,7a,12a,24~tetrahydroxycoprostanic acid 897-903.
by rat liver. Biochim. Biophys. Acta. 104 305-307. 64. Salen, G., S. Shefer, F. W. Cheng, B. Dayal, A. K. Batta,
48. Gustafsson, J. 1980. Biosynthesis of cholic acid in rat and G. S. Tint. 1979. Cholic acid biosynthesis: the en-
liver: formation of cholic acid from 3a,7a,12a- zymatic defect in cerebrotendinous xanthomatosis. J.
trihydroxy- and 3a,7a,12a,24tetrahydroxy-5pcholes- Clin. Invest. 6 3 38-44.
tanoic acids. Lipids. 1 5 113-121. 65. Duane, W., I. Bjbrkhem, J. N. Hamilton, and S. M.
49. Pedersen, J. I., and J. Gustafsson. 1980. Conversion of Mueller. 1988. Quantitative importance of the 25-
3a,7a,l2a-trihydroxy-5~holestanoicacid into cholic hydroxylase pathway for bile acid biosynthesis in the

Downloaded from www.jlr.org by guest, on May 14, 2019


acid by rat liver peroxisomes. EEBS Lett. 121: 345-348. rat. Hepatoha. 8 613-618.
50. Kase, F., I. Bjijrkhem, and J. I. Pedersen. 1983. Forma- 66. Duane, W. C., P. A. Pooler, and J. N. Hamilton. 1981.
tion of cholic acid from 3a,7a,12a-trihydroxy-5fl-choles- Bile acid synthesis in man. In vivo activity of the 25-
tanoic acid by rat liver peroxisomes. J. Lipid Res. 2 4 hydroxylase pathway. J. Clin. Invest. 8 2 82-85.
1560-1 567. 67. Mitropoulos, K. A., M. D. Avery, N. B. Myant, and G. F.
51. Prydz, IC, B. F. Kase, I. Bjbrkhem, and J. I. Pedersen. Gibbons. 1972. The formation of cholest-5ene-3P,26
1986. Formation of chenodeoxycholic acid from 3a,7a- diol as an intermediate in the conversion of cholesterol
dihydroxy-5pcholestanoicacid by rat liver peroxisomes. into bile acids by liver mitochondria. Biochem. J. 130
J. Lipid Res. 27: 622-628. 363-371.
52. Bjijrkhem, I., B. F. Kase, and J. I. Pedersen. 1984. 68. Mitropoulos, K. A., and N. B. Myant. 1967. The forma-
Mechanism of peroxisomal 24hydroxylation of tion of lithocholic acid, chenodeoxycholic acid and p
3a,7a,l2a-trihydroxy-5~holestanoic acid in rat liver. muricholic acid from cholesterol incubated with rat
Biochim. Biophys. Acta. 796: 142-145. liver mitochondria. Biochem.J. 103 472-479.
53. Ostlund-Farrants, A. R, I. Bjbrkhem, and J-I. Pedersen. 69. Swell, L., J. Gustafsson, C. C. Schwartz, L. G. Halloran,
1989. Identification of 3a,7a,l2a-trihydroxy-5~holest- H. Danielsson, and Z. R. Vlahcevic. 1980. An in vivo
24-enoic acid as an intermediate in the peroxisomal evaluation of the quantitative significance of several
conversion of 3t~,7a,l2a-trihydroxy-5~holestanoic potential pathways to cholic and chenodeoxycholic
acid to cholic acid. Biochim. Biophys. Acta. 1002: 198- acids from cholesterol in man.J. Lipid Res. 21: 455-466.
202. 70. Bjbrkhem, I., 0. Fausa, G. Hopen, H. Oftebro, J. I.
54. Prydz, R,B. F. Kase, I. Bjbrkhem, and J. I. Pedersen. Pedersen, and S. Skrede. 1983. Role of the 2 6
1988. Subcellular localization of 3a,7adihydroxy- and hydroxylase in the biosynthesis of bile acids in the nor-
3a,7a,12a-trihydroxy-5~cholestanoyl-CoA-ligase (s) in mal state and in CTX.J. Clin. Invest. 71: 142-148.
rat liver. J. Lipid Res. 29: 997-1004. 71. Bjbrkhem, I., H. Oftebro, S. Skrede, and J. I. Pedersen.
55. Pedersen, J. I., E. Hvattum, T. Flatabo, and I. Bjbrkhem. 1981. Assay of intermediates in bile acid biosynthesis
1988. Clofibrate does not induce peroxisomal using isotope dilution-mass spectrometry: hepatic
3a,7a,12a-trihydroxy-5Pcholestanoyl CoA oxidation in levels in the normal state and in cerebrotendinous xan-
rat liver. Evidence that this reaction is catalyzed by an thomatosis.J. Lipid Res. 2 2 191-200.
enzyme different from that of peroxisomal acyl-CoA 72. Axelson, M., B. Mbrk, and G. T. Everson. 1991. Bile
oxidation, Biochem. Znt. 17: 163-169. acid synthesis in cultured human hepatoblastoma cells.
56. Farrants-Ostlund, A. R , I. BjGrkhem, and J. I. Pedersen. J. Biol. Chem. 266: 17770-17777.
1990. Differential induction of peroxisomal oxidation 73. Axelson, M., and J. Sjbvall. 1990. Potential bile acid
of palmitic acid and 3a,7a,12a-trihydroxy-5~holes- precursors in plasma, possible indicators of biosynthetic
tanoic acid in rat liver. Biochim. Biophys. Acta. 1046: 173- pathways to cholic and chenodeoxycholic acid in man.
177. J. Steroid Biochem. 3 6 631-640.
57. Schepers, L., P. P. van Veldhoven, M. Casteels, H. J. Eys- 74. Krisans, S. IC, S. L. Thompson, L. A. Pena, E. Kok, and
sen, and G. P. Mannaerts. 1990. Presence of three acyl- N. B. Javitt. 1985. Bile acid synthesis in rat liver
CoA oxidases in rat liver peroxis0mes.J. Biol. Chem. 265 peroxisomes: metabolism of 26-hydroxycholesterol to
5242-5246. 3~hydroxy-5-cholenoicacid.J. Lipid Res. 2 6 1324-1332.
58. Gengenbacher, T., W. Gerok, U. Gise, W. W. Just, and 75. Anderson, IL E., E. Kok, and N. BrJavitt. 1972. Bile acid
G. Kurz. 1991. Peroxisomal proteins involved in bile salt synthesis in man: metabolism of 7a-hydroxycholesterol-
biosynthesis. In Bile acids as Therapeutic Agents. 14C and 26hydro~ycholesterol-~H. J. Clin. Invest. 51:
Kluwer Academic Publishers, Norwell, MA. 63-76. 112-1 17.

470 lournal of finid Research Volume 33. 1992


.#
76. Axelson, M., B. MBrk, and J. SjBvall. 1991. Studies on 91. Skrede, S., M. S. Buchmann, and I. Bjdrkhem. 1988.
biosynthetic pathways to cholic and chenodeoxycholic Hepatic 7adehydroxylation of bile acid intermediates,
acids in humans. In Bile acids as Therapeutic Agents. and its significance for the pathogenesis of cerebroten-
Falk Symposium no. 58. Kluwer Academic Publishing dinous xanthomatosis. J. Lipid Res. 29: 157-164.
Co., Norwell, MA. 53-62. 92. Kandutsch, A. A. 1963. Metabolism of cholesta4,7dien-
77. Axelson, M., B. MBrk, and J. SjGvall. 1988. Occurrence %ne and cholesta4,6dien-3-one by mouse liver micro-
of 3~hydroxy-5-cholestenoic acid, 3j3,7adihydroxy-5- somes. J. Lipid Res. 4 179-187.
cholestenoic acid and 7a-hydroxy-3-ox&holestenoic 93. Lazarow, P. B., and H. Moser. 1989. Disorders of
acid as normal constituents in human blood. J. Lipid peroxisome biogenesis. In The Metabolic Basis of In-
Res. 29: 629-641. herited Disease. Chapter 57. C. R. Scriver, A. L.
78. Javitt, N. B., E. Kok, B. Cohen, and S. Bursteon. 1982. Beaudet, W. S. Sly, and D. Valle, editors, McGraw-Hill
Cerebrotendinous xanthomatosis: reduced serum 26- Book Co., New York. 1479-1509.
hydroxycholesterol. J. Lipid Res. 23: 627-630. 94. Pedersen, J. I., B. F. Kase, K. Prydz, and 1. Bjdrkhem.
79. Princen, H. M. G., P. Meijer, B. G. Wolthers, R. J. Vonk, 1987. Role of peroxisomes in bile acid formation.
and F. Kuipers. 1991. Cyclosporin A blocks bile acid Preceedings of the X International Bile Acid Meeting,
synthesis in cultured hepatocytes by inhibition of Freiburg, June 1988. Kluwer Academic Publishers, Nor-
chenodeoxycholic acid and Pmuricholic acid synthesis. well, MA. 41-46.
B i o c h a . J. 2 7 5 501-505. 95. Kase, B. F., J. 1. Pedersen, B. Strandvik, and I.
80. Axelson, M., J. Skoda, J. SjBvall, A. Toll, and K. Wikvall. Bjdrkhem. 1985. In vivo and in vitro studies on forma-
1992. Cholesterol is converted to 7a-hydroxy-33x04 tion of bile acids in patients with Zellweger syndrome.
cholestenoic acid in liver mitochondria-evidence for a Evidence that peroxisomes are of importance in the
mitochondrial sterol 7a-hyroxylase. J. Biol. Chem. 267: normal biosynthesis of both cholic and cheno-
1701-1 704. deoxycholic acid. J. Clin. Invest. 76: 2393-2402.
81. BjBrkhem, I., and S. Skrede. 1989. Cerebrotendinous 96. Kase, B. F., I. Bjdrkhem, P. Higi, and J. I. Pedersen.
xanthomatosis and phytosterolemia. Chapter 51. In The 1985. Defective peroxisomal cleavage of the Cz;r-steroid

Downloaded from www.jlr.org by guest, on May 14, 2019


Metabolic Basis of Inherited Disease. C. R. Scriver, A. L. sidechain in cerebrohepatorenal syndrome of Zell-
Beaudet, W. S. Sly, and D. Valle, editors. McGraw-Hill weger. J. Clin. Invest. 75: 427-435.
Book, Co., New York. 1283-1302. 97. Kase, B. F., J. I. Pedersen, K. 0. Wathne, J. Gustafsson,
82. Setoguchi, T., G. Salen, G. S. Tint, and E. H. Mosbach. and I. BjBrkhem. 1991. Importance of peroxisomes in
1974. A biochemical abnormality in cerebrotendinous the formation of chenodeoxycholic acid in human
xanthomatosis. Impairment of bile acid biosynthesis a s liver. Metabolism of 3a,7adihydroxy-5~cholestanoic
sociated with incomplete degradation of the cholesterol acid in Zellweger syndrome. Pediatr. Res. 29: 64-69.
side chain.J. Clin. Invest. 531: 1393-1401. 98. Svoboda, J. A., M. J. Thompson, and W. E. Robbins.
83. Cali, J. J., GL. Hsieh, U. Francke, and D. W. Russel. 1967. Desmosterol, an intermediate in dealkylation of
1991. Mutations in the bile acid biosynthetic enzyme p-sitosterol in tobacco hornworm. 12ve Sci. 6: 395-404.
sterol 27-hydroxylase underlie cerebrotendinous xan- 99. Douglass, T. S., W. E. Connor, and D. S. Lin. 1981. The
thomatosis. J. Biol. Chem. 266 7779-7783. biosynthesis, absorption, apd origin of cholesterol and
84. Bjdrkhem, I., E. Reihntr, B. A. Angelin, S. Ewerth, J-E. plant sterols in the Florida land crab. J. Lipid Res. 22:
Akerlund, and K. Einarsson. 1987. On the possible use 961-970.
of the serum level of 7a-hydroxycholestero1 as a marker 100. Subbiah, M. T. R., and A. Kuksis. 1973. Differences in
for increased activity of the cholesterol 7a-hydroxylase metabolism of cholesterol and sitosterol following in-
in humans. J. Lipid Res. 28: 889-894. travenous injection in rats. Biochim. Biophys. Acta. 306:
85. Axelson, M., A. Aly, and J. SjBvall. 1988. Levels of 7a- 95-105.
hydroxykholesten-30ne in plasma reflect rates of bile 101. Kritchevsky, D., L. M. Davidsson, E. H. Mosbach, and B.
acid synthesis in man. FEBS Lett. 239: 324-328. I. Cohen. 1981. Identification of acid steroids in feces
86. Reihnkr, E., I. BjBrkhem, B. Angelin, S. Ewerth, and K. of monkeys fed p-sitosterol. Lipids. 16: 77-78.
Einarsson. 1989. Bile acid synthesis in humans: regula- 102. Salen, G., E. H. Ahrens, Jr., and S. M. Grundy. 1970.
tion of hepatic cholesterol 7a-hydroxylase activity. Metabolism of psitosterol in man. J. Clin. Invest. 4 9
Gastroenterology. 97: 1498-1505. 952-967.
87. Akerlund, J. E., and I. Bjdrkhem, and U. Andersson. 103. Muri-Boberg, K., K. Einarsson and I. Bjdrkhem. 1990.
1990. Studies on the regulation of cholesterol 7a- Apparent lack of conversion of sitosterol into C24-bile
hydroxylase and HMGCoA reductase in rat liver: ef- acids in humans.J. Lipid Res. 31: 1083-1088.
fects of lymphatic drainage and ligation of the lymph 104. Swell, L., and C. R. Treadwell. 1961. Metabolic fate of
duct. J. Lipid Res. 31: 2159-2166. injected ''C-phytosterols. h o c . Soc. Exp. Biol. Med. 108:
88. Duckworth, P. F., Z. R. Vlahcevic, E. J. Studer, E. C. 810-81 3.
Gurley, D. M. Heuman, Z. H. Beg, and P. B. Hylemon. 105. Skrede, B., I. Bjdrkhem, 0. Bergesen, H. J. Kayden, and
1991. Effect of hydrophobic bile acids on 3-hydroxy-3- S. Skrede. 1985. Studies on 5a-sitostanol: its presence in
methylglutaryl-CoA reductase activity and mRNA levels the serum of a patient with phytosterolemia and its
in the rat. J. Biol. Chem. 266: 9413-9418. biosynthesis from plant sterols in bile fistula rats.
89. Ballantyne, C. M., G. L. Vega, C. East, G. Richards, and Biochim. Biophys. Acta. 836: 368-375.
S. M. Grundy. 1987. Lowdensity lipoprotein metabo- 106. Muri-Boberg, K., E. Lund, J. Olund, and I. Bjdrkhem.
lism in cerebrotendinous xanthomatosis. Metabolism. 36: 1990. Formation of C-21 bile acids from plant sterols in
270-276. the rat. J. Biol. Chem. 265: 7967-7975.
90. Salen, G., T. W. Meriwether, and G. Nicolau. 1975. 107. Lund, E., K. Muri-Boberg, S. Bystrom, K. Carlstrdm, J.
Chenodeoxycholic acid inhibits increased cholesterol Olund, and I. Bjdrkhem. 1991. Formation of novel C21-
and cholestanol synthesis in patients with cerebroten- bile acids from cholesterol in the rat. J. Biol. Chem. 266:
dinous xanthomatosis. Biochem. Med. 14: 57-74. 4929-4937.

B j i i r k h Mechanism of degradation of steroid side chain m formation of bile acids 471

Вам также может понравиться